1
|
Wang Z, Modave E, Delfini M, Appeltans I, Boeckxstaens G, Stakenborg N. Large Peritoneal Macrophages Play No Role in the Pathogenesis of Postoperative Ileus Induced by Intestinal Manipulation. Neurogastroenterol Motil 2025; 37:e14997. [PMID: 39777769 DOI: 10.1111/nmo.14997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/16/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025]
Abstract
INTRODUCTION Postoperative ileus (POI) is an iatrogenic disorder marked by temporary impaired gastrointestinal (GI) motility post-abdominal surgery. Surgical handling of the intestine activates resident macrophages (Mfs), leading to inflammatory cytokine release and leukocyte recruitment into the muscularis, which compromises intestinal contractility. The mechanisms behind this activation are unclear. Recent studies suggest peritoneal Mfs, particularly large peritoneal macrophages (LPMs), might play a role in sterile intestinal inflammation by rapidly recruiting to the serosal layer of the gut and aiding in tissue damage resolution. METHODS To identify immune cells involved in the early phase of POI, single-cell RNA sequencing (scRNA-seq) was conducted. The migration of LPMs post-surgery was studied using adoptive transfer techniques. LPMs were depleted via intraperitoneal injection of clodronate liposomes. Subsequently, flow cytometry, quantitative PCR (qPCR), and immunofluorescence were performed to assess the impact of LPM depletion and analyze cell populations and inflammatory effects. RESULTS (1) Intestinal manipulation (IM) leads to the accumulation of monocytes, neutrophils, mature Mfs, CD8+ T cells, and LPMs within 2 h post-surgery. (2) Heparin treatment does not affect gut transit or reduce IL-6, IL-1a, and IL-1b expression in the early phase of POI. (3) Depletion of LPMs via clodronate liposome does not prevent monocyte, neutrophil, and Mfs infiltration in the muscularis externa, nor does it improve gut transit or reduce cytokine expression. (4) LPMs migrate to the serosa after IM but do not enter the muscularis externa. CONCLUSION AND INFERENCES LPMs adhere to the intestinal serosa following intestinal manipulation but do not migrate into the intestinal muscularis or participate in the inflammatory response and delayed transit. Consequently, LPMs are not involved in the pathogenesis of POI.
Collapse
Affiliation(s)
- Zheng Wang
- Center for Intestinal Neuro-Immune Interactions, Translational Research Center for GI Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Elodie Modave
- Center for Intestinal Neuro-Immune Interactions, Translational Research Center for GI Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Marcello Delfini
- Center for Intestinal Neuro-Immune Interactions, Translational Research Center for GI Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Iris Appeltans
- Center for Intestinal Neuro-Immune Interactions, Translational Research Center for GI Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Guy Boeckxstaens
- Center for Intestinal Neuro-Immune Interactions, Translational Research Center for GI Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Nathalie Stakenborg
- Center for Intestinal Neuro-Immune Interactions, Translational Research Center for GI Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| |
Collapse
|
2
|
Wang Z, Stakenborg N, Boeckxstaens G. Postoperative ileus-Immune mechanisms and potential therapeutic interventions. Neurogastroenterol Motil 2024:e14951. [PMID: 39462772 DOI: 10.1111/nmo.14951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 09/23/2024] [Accepted: 10/16/2024] [Indexed: 10/29/2024]
Abstract
BACKGROUND Postoperative ileus (POI) is a condition marked by a temporary suppression of gastrointestinal motility following abdominal surgery. The mechanism of POI encompasses various factors and is characterized by two phases: the early neurogenic phase involving both adrenergic and non-adrenergic neural pathways; the later immune-mediated phase is characterized by a sterile inflammatory response that lasts several days. Activation of muscularis macrophages triggers a sterile inflammatory process that results in dysfunction of the enteric nervous system (ENS) and a reversible inhibition of gastrointestinal motility. PURPOSE In this minireview, recent insights in the pathophysiological mechanisms underlying POI and potential new therapeutic strategies are described.
Collapse
Affiliation(s)
- Zheng Wang
- Center of Intestinal Neuro-Immune Interactions, Translational Research Center for GI Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing, KU Leuven-University of Leuven, Leuven, Belgium
| | - Nathalie Stakenborg
- Center of Intestinal Neuro-Immune Interactions, Translational Research Center for GI Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing, KU Leuven-University of Leuven, Leuven, Belgium
| | - Guy Boeckxstaens
- Center of Intestinal Neuro-Immune Interactions, Translational Research Center for GI Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing, KU Leuven-University of Leuven, Leuven, Belgium
| |
Collapse
|
3
|
Kong Q, Chen LM, Liu CY, Li W, Yin PH. The effect of acupuncture on gastrointestinal recovery after abdominal surgery: a narrative review from clinical trials. Int J Surg 2024; 110:5713-5721. [PMID: 38759698 PMCID: PMC11392097 DOI: 10.1097/js9.0000000000001641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 05/07/2024] [Indexed: 05/19/2024]
Abstract
Abdominal surgery is a critical surgery, with more and more attention being paid to postoperative life quality and associated complications in recent years. Among these complications, postoperative gastrointestinal dysfunction is the most common complication of abdominal surgery. Acupuncture therapy is a treatment approach based on the Traditional Chinese Medicine theory, and its feasibility in aiding gastrointestinal recovery after abdominal surgery is supported by both Traditional Chinese Medicine theory and animal experiments. A lot of clinical research has been conducted to evaluate its efficacy, albeit with limitations, and at preliminary stages. Moreover, intervention timing, acupoint selection, and patient benefits should also be considered in clinical practices. This article summarizes the progress of clinical research on acupuncture therapy in gastrointestinal recovery after abdominal surgery and discusses related issues and operations, with the aim to provide new insights and prospects for the incorporation of acupuncture into the Enhanced Recovery After Surgery protocol.
Collapse
Affiliation(s)
- Qi Kong
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li-Ming Chen
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chu-Yu Liu
- School of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Li
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Pei-Hao Yin
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
4
|
Wu XD, Yan HJ, Xu YM, Zhao SY, Zhang XD, Lv L, Zhu KL. Effect and mechanism of needleless transcutaneous neuromodulation on gastrointestinal function after pancreaticoduodenectomy. Scand J Gastroenterol 2024; 59:133-141. [PMID: 37752679 DOI: 10.1080/00365521.2023.2261060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/11/2023] [Accepted: 09/14/2023] [Indexed: 09/28/2023]
Abstract
BACKGROUND Gastrointestinal motility disorders tend to develop after pancreaticoduodenectomy (PD). The objectives of this study were: (1) to investigate the impact of needleless transcutaneous neuromodulation (TN) on the postoperative recuperation following pancreaticoduodenectomy (PD), and (2) to explore the underlying mechanisms by which TN facilitates the recovery of gastrointestinal function after PD. METHODS A total of 41 patients scheduled for PD were randomized into two groups: the TN group (n = 21) and the Sham-TN group (n = 20). TN was performed at acupoints ST-36 and PC-6 twice daily for 1 h from the postoperative day 1 (POD1) to day 7. Sham-TN was performed at non-acupoints. Subsequent assessments incorporated both heart rate variation and dynamic electrogastrography to quantify alterations in vagal activity (HF) and gastric pacing activity. RESULTS 1)TN significantly decreased the duration of the first passage of flatus (p < 0.001) and defecation (p < 0.01) as well as the time required to resume diet (p < 0.001) when compared to sham-TN;2)Compared with sham-TN, TN increased the proportion of regular gastric pacing activity (p < 0.01);3) From POD1 to POD7, there was a discernible augmentation in HF induced by TN stimulation(p < 0.01);4) TN significantly decreased serum IL-6 levels from POD1 to POD7 (p < 0.001);5) TN was an independent predictor of shortened hospital stay(β = - 0.349, p = 0.035). CONCLUSION Needleless TN accelerates the recovery of gastrointestinal function and reduces the risk of delayed gastric emptying in patients after PD by enhancing vagal activity and controlling the inflammatory response.
Collapse
Affiliation(s)
- Xu-Dong Wu
- Hepatopancreatobiliary Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo University, Ningbo, China
| | - Huan-Jun Yan
- Hepatopancreatobiliary Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo University, Ningbo, China
| | - Yue-Mei Xu
- Gastroenterology, The Affiliated People's Hospital of Ningbo University, Ningbo University, Ningbo, China
| | - Shuang-Ying Zhao
- Hepatopancreatobiliary Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo University, Ningbo, China
| | - Xiao-Dong Zhang
- Hepatopancreatobiliary Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo University, Ningbo, China
| | - Li- Lv
- Department of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Ke-Lei Zhu
- Hepatopancreatobiliary Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo University, Ningbo, China
| |
Collapse
|
5
|
Yuan PQ, Wu SV, Wang L, Taché Y. The ghrelin agonist, HM01 activates central vagal and enteric cholinergic neurons and reverses gastric inflammatory and ileus responses in rats. Neurogastroenterol Motil 2023; 35:e14561. [PMID: 36942655 DOI: 10.1111/nmo.14561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 02/09/2023] [Accepted: 02/24/2023] [Indexed: 03/23/2023]
Abstract
BACKGROUND Electrical vagal stimulation alleviates abdominal surgery (AS)-induced intestinal inflammation. Ghrelin receptors (GHS-Rs) are expressed in the brain and peripheral tissues. We investigated the influence of HM01, an orally active ghrelin agonist crossing the blood-brain barrier, on AS-induced gastric inflammation and emptying (GE) in rats. METHODS HM01 (6 mg/kg) or saline pretreatment was administered per orally (po) or intraperitoneally (ip). We assessed GE, gastric cytokine mRNA, and Fos positive cells in the dorsal motor nucleus of the vagus (DMN) and gastric corpus myenteric plexus (MP) in sham (anesthesia alone) and AS groups. The transcripts of GHS-R1 variants were determined in the medulla oblongata and gastric corpus of naïve rats. KEY RESULTS In vehicle pretreated rats, HM01 (ip) significantly increased the number of Fos immunoreactive cells in the MP and DMN in 55% and 52% of cholinergic neurons respectively. Hexamethonium did not modify HM01-induced Fos expression in the DMN while reducing it in the MP by 2-fold with values still significantly higher than that in control groups. AS upregulated gastric IL-1β and TNFα expression and inhibited GE by 66.6%. HM01 (po) abolished AS-induced gastric ileus and increased cytokine expression and elevated IL-10 by 4.0-fold versus vehicle/sham. GHS-R1a mRNA level was 5.4-fold higher than the truncated GHS-R1b isoform in the brain medulla and 40-fold higher in the gastric submucosa/muscle layers than in the mucosa. CONCLUSIONS AND INFERENCE Peripheral HM0 activates central vagal and myenteric cholinergic pathways that may influence both central and peripheral targets to prevent AS-induced gastric inflammatory and ileus.
Collapse
Affiliation(s)
- Pu-Qing Yuan
- VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, CURE: Digestive Diseases Research Center (DDRC), Center for Neurobiology of Stress and Resilience (CNSR), University of California Los Angeles, Los Angeles, California, USA
| | - S Vincent Wu
- VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Lixin Wang
- VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, CURE: Digestive Diseases Research Center (DDRC), Center for Neurobiology of Stress and Resilience (CNSR), University of California Los Angeles, Los Angeles, California, USA
| | - Yvette Taché
- VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, CURE: Digestive Diseases Research Center (DDRC), Center for Neurobiology of Stress and Resilience (CNSR), University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
6
|
Buscail E, Deraison C. Postoperative Ileus: a Pharmacological Perspective. Br J Pharmacol 2022; 179:3283-3305. [PMID: 35048360 DOI: 10.1111/bph.15800] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 12/31/2021] [Accepted: 01/05/2022] [Indexed: 11/29/2022] Open
Abstract
Post-operative ileus (POI) is a frequent complication after abdominal surgery. The consequences of POI can be potentially serious such as bronchial inhalation or acute functional renal failure. Numerous advances in peri-operative management, particularly early rehabilitation, have made it possible to decrease POI. Despite this, the rate of prolonged POI ileus remains high and can be as high as 25% of patients in colorectal surgery. From a pathophysiological point of view, POI has two phases, an early neurological phase and a later inflammatory phase, to which we could add a "pharmacological" phase during which analgesic drugs, particularly opiates, play a central role. The aim of this review article is to describe the phases of the pathophysiology of POI, to analyse the pharmacological treatments currently available through published clinical trials and finally to discuss the different research areas for potential pharmacological targets.
Collapse
Affiliation(s)
- Etienne Buscail
- IRSD, INSERM, INRAE, ENVT, University of Toulouse, CHU Purpan (University Hospital Centre), Toulouse, France.,Department of digestive surgery, colorectal surgery unit, Toulouse University Hospital, Toulouse, France
| | - Céline Deraison
- IRSD, INSERM, INRAE, ENVT, University of Toulouse, CHU Purpan (University Hospital Centre), Toulouse, France
| |
Collapse
|
7
|
Hellstrom EA, Ziegler AL, Blikslager AT. Postoperative Ileus: Comparative Pathophysiology and Future Therapies. Front Vet Sci 2021; 8:714800. [PMID: 34589533 PMCID: PMC8473635 DOI: 10.3389/fvets.2021.714800] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/19/2021] [Indexed: 12/11/2022] Open
Abstract
Postoperative ileus (POI), a decrease in gastrointestinal motility after surgery, is an important problem facing human and veterinary patients. 37.5% of horses that develop POI following small intestinal (SI) resection will not survive to discharge. The two major components of POI pathophysiology are a neurogenic phase which is then propagated by an inflammatory phase. Perioperative care has been implicated, namely the use of opioid therapy, inappropriate fluid therapy and electrolyte imbalances. Current therapy for POI variably includes an early return to feeding to induce physiological motility, reducing the inflammatory response with agents such as non-steroidal anti-inflammatory drugs (NSAIDs), and use of prokinetic therapy such as lidocaine. However, optimal management of POI remains controversial. Further understanding of the roles of the gastrointestinal microbiota, intestinal barrier function, the post-surgical inflammatory response, as well as enteric glial cells, a component of the enteric nervous system, in modulating postoperative gastrointestinal motility and the pathogenesis of POI may provide future targets for prevention and/or therapy of POI.
Collapse
Affiliation(s)
| | | | - Anthony T. Blikslager
- Department of Clinical Sciences, North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
8
|
Zhang B, Zhu K, Hu P, Xu F, Zhu L, Chen JDZ. Needleless Transcutaneous Neuromodulation Accelerates Postoperative Recovery Mediated via Autonomic and Immuno‐Cytokine Mechanisms in Patients With Cholecystolithiasis. Neuromodulation 2018; 22:546-554. [DOI: 10.1111/ner.12856] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Bo Zhang
- Department of GastroenterologyChangzheng Hospital, Second Military Medical University Shanghai China
- Ningbo Pace Translational Medical Research Center, Ningbo Zhejiang China
| | - Kelei Zhu
- Department of Hepatobiliary SurgeryYinzhou Hospital Ningbo China
| | - Pingping Hu
- Division of GastroenterologyYinzhou Hospital Ningbo China
| | - Feng Xu
- Division of GastroenterologyYinzhou Hospital Ningbo China
| | - Liang Zhu
- Department of GastroenterologyChangzheng Hospital, Second Military Medical University Shanghai China
| | - Jiande D. Z. Chen
- Ningbo Pace Translational Medical Research Center, Ningbo Zhejiang China
- Division of Gastroenterology and HepatologyJohns Hopkins Center for Neurogastroenterology Baltimore MD USA
| |
Collapse
|
9
|
Zhang B, Xu F, Hu P, Zhang M, Tong K, Ma G, Xu Y, Zhu L, Chen JDZ. Needleless Transcutaneous Electrical Acustimulation: A Pilot Study Evaluating Improvement in Post-Operative Recovery. Am J Gastroenterol 2018; 113:1026-1035. [PMID: 29925916 DOI: 10.1038/s41395-018-0156-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 04/30/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Functional gastrointestinal disturbance occurs after abdominal surgeries and could last for an extended period of time in some cases. This study was designed (1) to evaluate the effects of needleless transcutaneous electrical acustimulation (TEA) on postoperative recovery, and (2) to investigate the mechanisms involving autonomic function in postoperative patients after removal of gastrointestinal cancers. METHODS Forty-two patients (33 male, age: 69.5 ± 1.5 years) scheduled for abdominal surgical removal of gastrointestinal cancers were randomized to TEA (n = 21) and sham-TEA (n = 21). TEA was performed via acupoints ST36 and PC6 1 h twice daily from the postoperative day (POD) 1 to day 3. Sham-TEA was performed at non-acupoints. RESULTS (1) TEA improved major postoperative symptoms by about 30%, including a reduction in time to defecation by 31.7% (P < 0.01 vs. sham-TEA), time to first flatus by 35.9% (P < 0.001), time to ambulation by 42.8% (P < 0.01), time to resuming diet by 26.5% (P < 0.01) and hospital stay by 30% (P < 0.05) as well as pain score by 50% (P < 0.01). (2) TEA significantly increased vagal activity (P < 0.001) and decreased sympathetic activity on POD 4 (P < 0.001) compared with POD 1 as well as the serum level of NE (P < 0.05). (3) The vagal activity, high frequency assessed from the spectral analysis of heart rate variability, was negatively correlated with time to resuming diet, whereas the sympathetic measurement, serum norepinephrine was positively correlated with time to resuming diet and time to flatus. (4) TEA but not sham-TEA decreased TNF-α by 17.4% from POD 1 to POD 4. (5) TEA was an independent predictor of a shorter hospital stay. CONCLUSIONS Needleless TEA improves major postoperative symptoms by enhancing vagal and suppressing sympathetic activities.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Gastroenterology, Changzheng Hospital affiliated to Second Military Medical University, Shanghai, 200003, China. Ningbo Pace Translational Medical Research Center, Ningbo, Zhejiang, China. Division of Gastroenterology, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China. Gastroenterology Surgical Department, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China. Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China. Division of Gastroenterology and Hepatology, Johns Hopkins Center for Neurogastroenterology, Baltimore, MD 21224, USA. These authors contributed equally: Bo Zhang, Feng Xu.,Department of Gastroenterology, Changzheng Hospital affiliated to Second Military Medical University, Shanghai, 200003, China. Ningbo Pace Translational Medical Research Center, Ningbo, Zhejiang, China. Division of Gastroenterology, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China. Gastroenterology Surgical Department, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China. Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China. Division of Gastroenterology and Hepatology, Johns Hopkins Center for Neurogastroenterology, Baltimore, MD 21224, USA. These authors contributed equally: Bo Zhang, Feng Xu
| | - Feng Xu
- Department of Gastroenterology, Changzheng Hospital affiliated to Second Military Medical University, Shanghai, 200003, China. Ningbo Pace Translational Medical Research Center, Ningbo, Zhejiang, China. Division of Gastroenterology, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China. Gastroenterology Surgical Department, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China. Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China. Division of Gastroenterology and Hepatology, Johns Hopkins Center for Neurogastroenterology, Baltimore, MD 21224, USA. These authors contributed equally: Bo Zhang, Feng Xu
| | - Pingping Hu
- Department of Gastroenterology, Changzheng Hospital affiliated to Second Military Medical University, Shanghai, 200003, China. Ningbo Pace Translational Medical Research Center, Ningbo, Zhejiang, China. Division of Gastroenterology, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China. Gastroenterology Surgical Department, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China. Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China. Division of Gastroenterology and Hepatology, Johns Hopkins Center for Neurogastroenterology, Baltimore, MD 21224, USA. These authors contributed equally: Bo Zhang, Feng Xu
| | - Mingyuan Zhang
- Department of Gastroenterology, Changzheng Hospital affiliated to Second Military Medical University, Shanghai, 200003, China. Ningbo Pace Translational Medical Research Center, Ningbo, Zhejiang, China. Division of Gastroenterology, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China. Gastroenterology Surgical Department, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China. Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China. Division of Gastroenterology and Hepatology, Johns Hopkins Center for Neurogastroenterology, Baltimore, MD 21224, USA. These authors contributed equally: Bo Zhang, Feng Xu
| | - Kehui Tong
- Department of Gastroenterology, Changzheng Hospital affiliated to Second Military Medical University, Shanghai, 200003, China. Ningbo Pace Translational Medical Research Center, Ningbo, Zhejiang, China. Division of Gastroenterology, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China. Gastroenterology Surgical Department, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China. Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China. Division of Gastroenterology and Hepatology, Johns Hopkins Center for Neurogastroenterology, Baltimore, MD 21224, USA. These authors contributed equally: Bo Zhang, Feng Xu
| | - Gang Ma
- Department of Gastroenterology, Changzheng Hospital affiliated to Second Military Medical University, Shanghai, 200003, China. Ningbo Pace Translational Medical Research Center, Ningbo, Zhejiang, China. Division of Gastroenterology, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China. Gastroenterology Surgical Department, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China. Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China. Division of Gastroenterology and Hepatology, Johns Hopkins Center for Neurogastroenterology, Baltimore, MD 21224, USA. These authors contributed equally: Bo Zhang, Feng Xu.,Department of Gastroenterology, Changzheng Hospital affiliated to Second Military Medical University, Shanghai, 200003, China. Ningbo Pace Translational Medical Research Center, Ningbo, Zhejiang, China. Division of Gastroenterology, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China. Gastroenterology Surgical Department, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China. Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China. Division of Gastroenterology and Hepatology, Johns Hopkins Center for Neurogastroenterology, Baltimore, MD 21224, USA. These authors contributed equally: Bo Zhang, Feng Xu
| | - Yuemei Xu
- Department of Gastroenterology, Changzheng Hospital affiliated to Second Military Medical University, Shanghai, 200003, China. Ningbo Pace Translational Medical Research Center, Ningbo, Zhejiang, China. Division of Gastroenterology, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China. Gastroenterology Surgical Department, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China. Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China. Division of Gastroenterology and Hepatology, Johns Hopkins Center for Neurogastroenterology, Baltimore, MD 21224, USA. These authors contributed equally: Bo Zhang, Feng Xu
| | - Liang Zhu
- Department of Gastroenterology, Changzheng Hospital affiliated to Second Military Medical University, Shanghai, 200003, China. Ningbo Pace Translational Medical Research Center, Ningbo, Zhejiang, China. Division of Gastroenterology, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China. Gastroenterology Surgical Department, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China. Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China. Division of Gastroenterology and Hepatology, Johns Hopkins Center for Neurogastroenterology, Baltimore, MD 21224, USA. These authors contributed equally: Bo Zhang, Feng Xu
| | - Jiande D Z Chen
- Department of Gastroenterology, Changzheng Hospital affiliated to Second Military Medical University, Shanghai, 200003, China. Ningbo Pace Translational Medical Research Center, Ningbo, Zhejiang, China. Division of Gastroenterology, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China. Gastroenterology Surgical Department, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China. Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China. Division of Gastroenterology and Hepatology, Johns Hopkins Center for Neurogastroenterology, Baltimore, MD 21224, USA. These authors contributed equally: Bo Zhang, Feng Xu.,Department of Gastroenterology, Changzheng Hospital affiliated to Second Military Medical University, Shanghai, 200003, China. Ningbo Pace Translational Medical Research Center, Ningbo, Zhejiang, China. Division of Gastroenterology, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China. Gastroenterology Surgical Department, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China. Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China. Division of Gastroenterology and Hepatology, Johns Hopkins Center for Neurogastroenterology, Baltimore, MD 21224, USA. These authors contributed equally: Bo Zhang, Feng Xu
| |
Collapse
|
10
|
First experience of the Egyptian National Cancer Institute using the robot-assisted laparoscopic approach in radical hysterectomies for cervical cancer. J Egypt Natl Canc Inst 2018; 30:61-67. [PMID: 29691094 DOI: 10.1016/j.jnci.2018.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 03/23/2018] [Accepted: 03/29/2018] [Indexed: 11/21/2022] Open
Abstract
AIM OF WORK Robotics in surgery led to an improvement of visualization, a better handling of tissues and better suturing. This study aimed to document the first experience of the Egyptian National Cancer Institute (NCI) using the robot-assisted laparoscopic approach in radical hysterectomies for cervical cancer and to highlight observed advantages, disadvantages, morbidity and oncological outcomes. PATIENTS AND METHODS Data of cases that had either early cervical cancer (stage IB-IIA1 with a tumor ≥2 cm) or locally advanced cervical cancer (Stage IIA2-IIB after chemo-radiotherapy) were collected prospectively. Study patients underwent robotic radical hysterectomies for their cervical cancers at the NCI, Cairo University, between January 1, 2015 and December 31, 2016. For each patient, duration of surgery, amount of blood loss, and intra-operative complications were recorded. Similarly, the duration of postoperative hospital-stay, analgesia used and post-operative gastrointestinal recovery were documented. Pathological assessment of safety margins and the lymph nodes number yield were also assessed. RESULTS Twenty patients underwent robotic radical hysterectomy during the study period. Twelve cases had early cervical cancer while 8 suffered locally advanced disease. The mean procedure time was 319 (range 240-560) minutes; the mean blood loss was 309 (range 150-600) ml. Three cases had bladder injuries during their procedures. The median hospital stay was 6 (range 4-10) days. One case had a positive margin. The median of lymph nodes yield number was 15 (range 10-25). Follow-up ranged 9-31 months, with only one case developing local recurrence. CONCLUSION Robotic radical hysterectomy is a feasible approach with a tolerable rate of complications.
Collapse
|
11
|
Lisowski ZM, Pirie RS, Blikslager AT, Lefebvre D, Hume DA, Hudson NPH. An update on equine post-operative ileus: Definitions, pathophysiology and management. Equine Vet J 2018; 50:292-303. [DOI: 10.1111/evj.12801] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 11/24/2017] [Indexed: 12/18/2022]
Affiliation(s)
- Z. M. Lisowski
- The Roslin Institute and Royal (Dick) School of Veterinary Studies; University of Edinburgh, Easter Bush; Midlothian UK
| | - R. S. Pirie
- The Roslin Institute and Royal (Dick) School of Veterinary Studies; University of Edinburgh, Easter Bush; Midlothian UK
| | - A. T. Blikslager
- Department of Clinical Sciences; College of Veterinary Medicine; North Carolina State University; Raleigh North Carolina USA
| | - D. Lefebvre
- The Roslin Institute and Royal (Dick) School of Veterinary Studies; University of Edinburgh, Easter Bush; Midlothian UK
| | - D. A. Hume
- The Roslin Institute and Royal (Dick) School of Veterinary Studies; University of Edinburgh, Easter Bush; Midlothian UK
- Mater Research; The University of Queensland; Woolloongabba Queensland Australia
| | - N. P. H. Hudson
- The Roslin Institute and Royal (Dick) School of Veterinary Studies; University of Edinburgh, Easter Bush; Midlothian UK
| |
Collapse
|
12
|
Stakenborg N, Gomez-Pinilla PJ, Boeckxstaens GE. Postoperative Ileus: Pathophysiology, Current Therapeutic Approaches. Handb Exp Pharmacol 2017; 239:39-57. [PMID: 27999957 DOI: 10.1007/164_2016_108] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Postoperative ileus, which develops after each abdominal surgical procedure, is an iatrogenic disorder characterized by a transient inhibition of gastrointestinal motility. Its pathophysiology is complex involving pharmacological (opioids, anesthetics), neural, and immune-mediated mechanisms. The early neural phase, triggered by activation of afferent nerves during the surgical procedure, is short lasting compared to the later inflammatory phase. The latter starts after 3-6 h and lasts several days, making it a more interesting target for treatment. Insight into the triggers and immune cells involved is of great importance for the development of new therapeutic strategies. In this chapter, the pathogenesis and the current therapeutic approaches to treat postoperative ileus are discussed.
Collapse
Affiliation(s)
- N Stakenborg
- Division of Gastroenterology and Hepatology, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Herestraat 49, Leuven, 3000, Belgium
| | - P J Gomez-Pinilla
- Division of Gastroenterology and Hepatology, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Herestraat 49, Leuven, 3000, Belgium
| | - G E Boeckxstaens
- Division of Gastroenterology and Hepatology, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Herestraat 49, Leuven, 3000, Belgium. .,Division of Gastroenterology and Hepatology, University Hospital Leuven, Herestraat 49, Leuven, 3000, Belgium.
| |
Collapse
|
13
|
Fang JF, Fang JQ, Shao XM, Du JY, Liang Y, Wang W, Liu Z. Electroacupuncture treatment partly promotes the recovery time of postoperative ileus by activating the vagus nerve but not regulating local inflammation. Sci Rep 2017; 7:39801. [PMID: 28051128 PMCID: PMC5209726 DOI: 10.1038/srep39801] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 11/29/2016] [Indexed: 12/11/2022] Open
Abstract
Postoperative ileus (POI) after abdominal surgery significantly lowers the life quality of patients and increase hospital costs. However, few treatment strategies have successfully shortened the duration of POI. Electroacupuncture (EA) is a modern way of administering acupuncture and widely used in various gastrointestinal (GI) diseases in the world. Here, we studied the effect of EA on POI and its underlying mechanisms. Intestinal manipulation resulted in significant delays of GI transit, colonic transit and gastric emptying. Surgery also up-regulated c-fos in nucleus of the solitary tract (NTS) and induced inflammation response in the small intestine. Further, operation and inhale anesthesia inhibited NTS neuron excitation duration for the whole observation time. EA administered at ST36 indeed shortened the recovery time of GI and colonic transit, and significantly increased the gastric emptying. EA also significantly activated the NTS neurons after operation. However, there was no anti-inflammation effect of EA during the whole experiment. Finally, atropine blocked the regulatory effect of EA on GI function, when it was injected after surgery, but not before surgery. Thus, the regulatory effect of EA on POI was mainly mediated by exciting NTS neurons to improve the GI tract transit function but not by activating cholinergic anti-inflammatory pathway.
Collapse
Affiliation(s)
- Jun-Fan Fang
- Department of Neurobiology &Acupuncture Research, the Third Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jian-Qiao Fang
- Department of Neurobiology &Acupuncture Research, the Third Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiao-Mei Shao
- Department of Neurobiology &Acupuncture Research, the Third Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jun-Ying Du
- Department of Neurobiology &Acupuncture Research, the Third Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yi Liang
- Department of Neurobiology &Acupuncture Research, the Third Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wen Wang
- Department of Neurobiology &Acupuncture Research, the Third Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhe Liu
- Department of Neurobiology &Acupuncture Research, the Third Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
14
|
Liang C, Wang K, Xu B, Yu Z. Electroacupuncture at acupoint ST 37(Shangjuxu) improves function of the enteric nervous system in a novel mouse constipation model. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 16:392. [PMID: 27756367 PMCID: PMC5070084 DOI: 10.1186/s12906-016-1377-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 10/13/2016] [Indexed: 02/07/2023]
Abstract
Background Electroacupuncture (EA) at acupoint ST 37 (Shangjuxu) has been used to alleviate gastrointestinal symptoms and improve gastrointestinal motility. However, the mechanisms by which EA affects the enteric nervous system (ENS) have scarcely been investigated. In this study, we investigated whether EA could improve ENS function. Methods A constipation model was established by gastric instillation of ice-cold saline daily for 14 days. The constipated mice were divided into two groups: the model group, which was not treated, and the EA group, which received EA at ST 37 at a frequency of 2–15 HZ and an amplitude of 1 mA for 15 min a day for 3 days. A further six mice were included as a non-constipated control group. After EA treatment, intestinal propulsion and defecation time were measured. Additionally, in jejunum, ileum and proximal colon myenteric plexus, the expressions of PGP9.5 and nNOS were measured by immunohistochemistry. Results The EA group demonstrated significant improvements in carbon propulsion rates and defecation time compared to model group (P < 0.05). In addition, after EA, the PGP9.5 and nNOS expression in jejunum, ileum and proximal colonic myenteric plexus was back to normal levels. Conclusion This study suggests that EA stimulation at ST 37 is capable of ameliorating intestinal motility dysfunction, and can partly restore enteric neuron function. The ENS can participate in changes in intestinal motility by affecting inhibitory neurons.
Collapse
|
15
|
Lies B, Beck K, Keppler J, Saur D, Groneberg D, Friebe A. Nitrergic signalling via interstitial cells of Cajal regulates motor activity in murine colon. J Physiol 2015; 593:4589-601. [PMID: 26227063 DOI: 10.1113/jp270511] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 07/16/2015] [Indexed: 12/11/2022] Open
Abstract
In the enteric nervous systems, NO is released from nitrergic neurons as a major inhibitory neurotransmitter. NO acts via NO-sensitive guanylyl cyclase (NO-GC), which is found in different gastrointestinal (GI) cell types including smooth muscle cells (SMCs) and interstitial cells of Cajal (ICC). The precise mechanism of nitrergic signalling through these two cell types to regulate colonic spontaneous contractions is not fully understood yet. In the present study we investigated the impact of endogenous and exogenous NO on colonic contractile motor activity using mice lacking nitric oxide-sensitive guanylyl cyclase (NO-GC) globally and specifically in SMCs and ICC. Longitudinal smooth muscle of proximal colon from wild-type (WT) and knockout (KO) mouse strains exhibited spontaneous contractile activity ex vivo. WT and smooth muscle-specific guanylyl cyclase knockout (SMC-GCKO) colon showed an arrhythmic contractile activity with varying amplitudes and frequencies. In contrast, colon from global and ICC-specific guanylyl cyclase knockout (ICC-GCKO) animals showed a regular contractile rhythm with constant duration and amplitude of the rhythmic contractions. Nerve blockade (tetrodotoxin) or specific blockade of NO signalling (L-NAME, ODQ) did not significantly affect contractions of GCKO and ICC-GCKO colon whereas the arrhythmic contractile patterns of WT and SMC-GCKO colon were transformed into uniform motor patterns. In contrast, the response to electric field-stimulated neuronal NO release was similar in SMC-GCKO and global GCKO. In conclusion, our results indicate that basal enteric NO release acts via myenteric ICC to influence the generation of spontaneous contractions whereas the effects of elevated endogenous NO are mediated by SMCs in the murine proximal colon.
Collapse
Affiliation(s)
- Barbara Lies
- Physiologisches Institut, Universität Würzburg, Würzburg, Germany
| | - Katharina Beck
- Physiologisches Institut, Universität Würzburg, Würzburg, Germany
| | - Jonas Keppler
- Physiologisches Institut, Universität Würzburg, Würzburg, Germany
| | - Dieter Saur
- II. Medizinische Klinik und Poliklinik, Technische Universität München, München, Germany
| | - Dieter Groneberg
- Physiologisches Institut, Universität Würzburg, Würzburg, Germany
| | - Andreas Friebe
- Physiologisches Institut, Universität Würzburg, Würzburg, Germany
| |
Collapse
|
16
|
Vather R, O'Grady G, Bissett IP, Dinning PG. Postoperative ileus: mechanisms and future directions for research. Clin Exp Pharmacol Physiol 2014; 41:358-70. [PMID: 24754527 DOI: 10.1111/1440-1681.12220] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 02/13/2014] [Accepted: 02/25/2014] [Indexed: 12/13/2022]
Abstract
Postoperative ileus (POI) is an abnormal pattern of gastrointestinal motility characterized by nausea, vomiting, abdominal distension and/or delayed passage of flatus or stool, which may occur following surgery. Postoperative ileus slows recovery, increases the risk of developing postoperative complications and confers a significant financial load on healthcare institutions. The aim of the present review is to provide a succinct overview of the clinical features and pathophysiological mechanisms of POI, with final comment on selected directions for future research.Terminology used when describing POI is inconsistent, with little differentiation made between the obligatory period of gut dysfunction seen after surgery ('normal POI') and the more clinically and pathologically significant entity of a 'prolonged POI'. Both normal and prolonged POI represent a fundamentally similar pathophysiological phenomenon. The aetiology of POI is postulated to be multifactorial, with principal mediators being inflammatory cell activation, autonomic dysfunction (both primarily and as part of the surgical stress response), agonism at gut opioid receptors, modulation of gastrointestinal hormone activity and electrolyte derangements. A final common pathway for these effectors is impaired contractility and motility and gut wall oedema. There are many potential directions for future research. In particular, there remains scope to accurately characterize the gastrointestinal dysfunction that underscores an ileus, development of an accurate risk stratification tool will facilitate early implementation of preventive measures and clinical appraisal of novel therapeutic strategies that target individual pathways in the pathogenesis of ileus warrant further investigation.
Collapse
Affiliation(s)
- Ryash Vather
- Department of Surgery, University of Auckland, Auckland, New Zealand
| | | | | | | |
Collapse
|
17
|
Gomez-Pinilla PJ, Binda MM, Lissens A, Di Giovangiulio M, van Bree SH, Nemethova A, Stakenborg N, Farro G, Bosmans G, Matteoli G, Deprest J, Boeckxstaens GE. Absence of intestinal inflammation and postoperative ileus in a mouse model of laparoscopic surgery. Neurogastroenterol Motil 2014; 26:1238-47. [PMID: 24966010 DOI: 10.1111/nmo.12376] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 05/08/2014] [Indexed: 02/08/2023]
Abstract
BACKGROUND Postoperative ileus (POI) is characterized by impaired gastrointestinal motility resulting from intestinal handling-associated inflammation. The introduction of laparoscopic surgery has dramatically reduced the duration of POI. However, it remains unclear to what extent this results in a reduction of intestinal inflammation. The aim of the present study is to compare the degree of intestinal inflammation and gastrointestinal transit following laparoscopic surgery and open abdominal surgery. METHODS Mice were subjected to laparoscopic surgery or laparotomy alone or, in combination with standardized intestinal manipulation of the small bowel (IM). Gastrointestinal transit and intestinal inflammation were assessed 24 h after surgery by the number of myeloperoxidase (MPO) positive cells and the level of cytokine expression. The recovery time and the degree of inflammation were also analyzed in patients subjected to colectomy under open conditions (laparotomy) or laparoscopic conditions. KEY RESULTS Mice undergoing IM by laparotomy (open IM), but not by laparoscopy (Lap IM) developed a significant delay in gastrointestinal transit compared to laparotomy or laparoscopy alone. In addition, there was significant intestinal inflammation only after open IM. Similarly, cytokine levels in peritoneal lavage fluid were lower while recovery time was faster in patients subjected to colectomy under laparoscopic conditions compared to open colectomy. CONCLUSIONS & INFERENCES Our data confirms that intestinal inflammation is underlying the delayed gastrointestinal transit observed after open surgery. Most importantly, we demonstrate that intestinal inflammation under laparoscopic conditions is significantly lower compared to open surgery, most likely explaining the faster recovery following laparoscopic surgery.
Collapse
Affiliation(s)
- Pedro J Gomez-Pinilla
- Translational Research in GastroIntestinal Disorders (TARGID) and Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Zhao J, Gao B, Zhang Y, Zheng B, Liu H, Cao JL. Effects of intrathecal opioids combined with low-dose naloxone on motilin and its receptor in a rat model of postoperative pain. Life Sci 2014; 103:88-94. [DOI: 10.1016/j.lfs.2014.03.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 03/12/2014] [Accepted: 03/31/2014] [Indexed: 12/28/2022]
|
19
|
Goetz B, Benhaqi P, Glatzle J, Müller MH, Schmitt SM, Brändli AW, Kreis ME, Kasparek MS. Changes in peptidergic neurotransmission during postoperative ileus in rat circular jejunal muscle. Neurogastroenterol Motil 2014; 26:397-409. [PMID: 24330008 DOI: 10.1111/nmo.12275] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 11/14/2013] [Indexed: 02/08/2023]
Abstract
BACKGROUND Our aim was to explore unknown changes in neurotransmission with vasoactive intestinal peptide (VIP) and Substance P (Sub P) during postoperative ileus (POI). METHODS Contractile activity of rat circular jejunal muscle strips was studied in five groups (n = 6/group): Naïve controls, sham controls 12 h and 3 days after laparotomy, and rats 12 h, 3 days after induction of POI. Dose-responses to VIP (10(-10) -10(-7) M), Sub P (3 × 10(-10) -3 × 10(-7) M), and electrical field stimulation (EFS, to study endogenous release of neurotransmitters) were studied with different antagonists. Intestinal transit, inflammatory cells and immunoreactivity for VIP and Sub P were investigated in the bowel wall and cellular Finkel osteo sarcoma expression was determined in vagal afferent and efferent nuclei of the brainstem. KEY RESULTS Postoperative ileus characterized by delayed intestinal transit and intramural inflammation was associated with an increased inhibitory effect of VIP on contractile activity. A biphasic impact was observed for Sub P with a decrease in its excitatory potential on contractility at 12 h, followed by a later increase 3 days postoperatively. Inhibitory response to EFS was increased, whereas the excitatory response decreased in ileus animals. VIP expression was increased in all postoperative animals while only animals 3 days after ileus induction showed increased Sub P expression in the myenteric plexus. These changes were associated with an activation of afferent but not efferent vagal nuclei in the brain stem. CONCLUSIONS & INFERENCES Specific, time-dependent changes in peptidergic neurotransmission with VIP and Sub P occur during POI that are associated with vagal afferent activation, but are independent of the activation of efferent vagal pathways.
Collapse
Affiliation(s)
- B Goetz
- Walter-Brendel-Center of Experimental Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Koscielny A, Engel D, Maurer J, Wehner S, Kurts C, Kalff JC. The role of lymphoid tissue in the attenuation of the postoperative ileus. Am J Physiol Gastrointest Liver Physiol 2013; 304:G401-12. [PMID: 23238935 DOI: 10.1152/ajpgi.00161.2012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Standardized intestinal manipulation (IM) leads to local bowel wall inflammation subsequently spreading over the entire gastrointestinal tract. Previously, we demonstrated that this so-called gastrointestinal field effect (FE) is immune-mediated. The aim of this study was to investigate the role of secondary lymphoid organs [mesenteric lymph nodes (MLN), gut-associated lymphoid tissue (GALT)] in IM-mediated FE by employing mice with deficient secondary lymphoid organs (aly/aly, MLN ex) or by administration of 2-amino-2-[2-(4-octylphenyl)ethyl]-1,3-propanediol (FTY720), an immunomodulating agent that inhibits emigration of lymphocytes out of lymphoid organs. Small bowel muscularis, and colonic muscularis from wild-type mice as control, from aly/aly mice, FTY720-treated mice (daily dose of 1.0 mg/kg mouse ip starting 3 days before surgical procedure), and wild-type mice that had undergone removal of mesenteric lymph nodes before IM (MLN ex mice) were obtained after selective IM of the jejunum or sham operation. FE was analyzed by measuring transit time of orally administered fluorescent dextran in the gastrointestinal tract [geometric center (GC) of fluorescent dextran], colonic transit time, infiltration of myeloperoxidase-positive cells, and circular smooth muscle contractility. Furthermore, mRNA levels of inflammatory cytokines [interleukin (IL)-6, tumor necrosis factor (TNF)-α, macrophage inflammatory protein (MIP)-1α] were determined by Taqman-PCR. We observed a significantly reduced upregulation of proinflammatory cytokines (IL-6, TNF-α, MIP-1α) in colonic muscularis of MLN ex mice, aly/aly mice, and FTY720-treated mice compared with wild-type mice. Contractility of circular muscularis strips of the colon but not the jejunum was significantly improved in aly/aly mice and FTY720-treated wild-type mice. Additionally, inflammation of the colon determined by the number of myeloperoxidase-positive cells and colonic transit time were significantly improved in aly/aly mice, FTY720-treated wild-type mice, and in MLN ex mice. In summary, lack of secondary lymphoid organs (MLN + GALT) in aly/aly mice or administration of FTY720 abrogated FE after IM as opposed to wild-type mice. These data demonstrate that secondary lymphoid organs are involved in the propagation of FE and postoperative ileus. FTY720 indirectly affects FE by inhibiting migration of activated T cells from the jejunum and adjacent secondary lymphoid organs to the colon. These findings support the crucial role of the adaptive immune system in FE, most likely by a sphyngosine 1-phosphate-dependent mechanism.
Collapse
Affiliation(s)
- A Koscielny
- Department of Surgery, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany.
| | | | | | | | | | | |
Collapse
|
21
|
Goetz B, Benhaqi P, Müller MH, Kreis ME, Kasparek MS. Changes in beta-adrenergic neurotransmission during postoperative ileus in rat circular jejunal muscle. Neurogastroenterol Motil 2013; 25:154-e84. [PMID: 23009554 DOI: 10.1111/nmo.12020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND To explore postoperative changes in β-adrenergic neurotransmission that participate in pathophysiology of postoperative ileus. METHODS Contractile activity of circular jejunal muscle strips was studied. Groups (n = 6/group) were: naïve controls, sham controls 1 and 7 days after laparotomy, and rats 12 h, 1, 3, and 7 days after laparotomy with standardized small bowel manipulation (postoperative ileus). Dose-responses to the β-agonist isoprenaline (3 × 10(-10) - 10(-7) mol L(-1)) were studied in presence/absence of tetrodotoxin (global neural blockade; 10(-6) mol L(-1) ), N6-(1-iminoethyl)-l-lysine (inhibition of inducible nitric oxide synthesis; 10(-4) mol L(-1)), nimesulide (cyclooxygenase-2 inhibition; 10(-5) mol L(-1)), or propranolol (β-blockade; 5 × 10(-6) mol L(-1)). Histochemistry for inflammatory cells and intestinal transit were studied. KEY RESULTS Intramural inflammation and delayed transit (postoperative ileus) occurred only in ileus groups. The inhibitory potential of isoprenaline decreased in all postoperative groups including sham (P < 0.05). Tetrodotoxin enhanced isoprenaline-induced inhibition in ileus and sham groups (P < 0.05). N6-(1-iminoethyl)-l-lysine and nimesulide decreased isoprenaline-induced inhibition in ileus groups 12 h, 1, and 7 days, and in sham controls 7 days postoperatively (P < 0.05). Propranolol prevented isoprenaline effects in all groups (P < 0.05). CONCLUSIONS & INFERENCES Inhibitory effects of isoprenaline on contractile activity were decreased for 7 days postoperatively. Changes in β-adrenergic neurotransmission do not induce postoperative ileus and appear to be caused by anesthesia and laparotomy rather than bowel manipulation.
Collapse
Affiliation(s)
- B Goetz
- Walter Brendel Center of Experimental Medicine, Munich, Germany
| | | | | | | | | |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW The pathophysiological riddle of the clinically important postoperative ileus (POI) has been solved more and more over the last decade. The POI is caused by inflammation and paralysis at the manipulated site propagating to the entire, unmanipulated gastrointestinal tract. Intestinal macrophages produce mediators that paralyze myocytes, but it is unclear how macrophages are activated, particularly those in unmanipulated areas. In addition to direct or neurally mediated activation of intestinal macrophages, a new immunologically mediated activation has been proposed. RECENT FINDINGS Recently, it has been shown that the surgical trauma induces interleukin-12 (IL-12) production by intestinal dendritic cells, which activates TH1-memory cells at the manipulated site. Those TH1-memory cells produce interferon-γ (IFN-γ). Those TH1 CCR9 cells also migrate to unmanipulated parts of the gastrointestinal tract. Their IFN-γ stimulates intestinal macrophages to produce nitirc oxide paralyzing myocytes leading to gastrointestinal hypomotility. SUMMARY The involvement of the adaptive (T-helper type 1 cell-mediated immune response) and of the innate (mast cells, intestinal macrophages) immune system in the pathophysiology of POI displays possible targets for objective monitoring and treatment of POI.
Collapse
|
23
|
STENGEL A, GOEBEL-STENGEL M, WANG L, LUCKEY A, HU E, RIVIER J, TACHÉ Y. Central administration of pan-somatostatin agonist ODT8-SST prevents abdominal surgery-induced inhibition of circulating ghrelin, food intake and gastric emptying in rats. Neurogastroenterol Motil 2011; 23:e294-308. [PMID: 21569179 PMCID: PMC3117963 DOI: 10.1111/j.1365-2982.2011.01721.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Activation of brain somatostatin receptors (sst(1-5) ) with the stable pan-sst(1-5) somatostatin agonist, ODT8-SST blocks acute stress and central corticotropin-releasing factor (CRF)-mediated activation of endocrine and adrenal sympathetic responses. Brain CRF signaling is involved in delaying gastric emptying (GE) immediately post surgery. We investigated whether activation of brain sst signaling pathways modulates surgical stress-induced inhibition of gastric emptying and food intake. METHODS Fasted rats were injected intracisternally (i.c.) with somatostatin agonists and underwent laparotomy and 1-min cecal palpation. Gastric emptying of a non-nutrient solution and circulating acyl and desacyl ghrelin levels were assessed 50min post surgery. Food intake was monitored for 24 h. KEY RESULTS The abdominal surgery-induced inhibition of GE (65%), food intake (73% at 2h) and plasma acyl ghrelin levels (67%) was completely prevented by ODT8-SST (1μg per rat, i.c.). The selective sst(5) agonist, BIM-23052 prevented surgery-induced delayed GE, whereas selective sst(1) , sst(2) , or sst(4) agonists had no effect. However, the selective sst(2) agonist, S-346-011 (1μg per rat, i.c.) counteracted the abdominal surgery-induced inhibition of acyl ghrelin and food intake but not the delayed GE. The ghrelin receptor antagonist, [D-Lys(3) ]-GHRP-6 (0.93mg kg(-1) , intraperitoneal, i.p.) blocked i.p. ghrelin-induced increased GE, while not influencing i.c. ODT8-SST-induced prevention of delayed GE and reduced food intake after surgery. CONCLUSIONS & INFERENCES ODT8-SST acts in the brain to prevent surgery-induced delayed GE likely via activating sst(5) . ODT8-SST and the sst(2) agonist prevent the abdominal surgery-induced decrease in food intake and plasma acyl ghrelin indicating dissociation between brain somatostatin signaling involved in preventing surgery-induced suppression of GE and feeding response.
Collapse
Affiliation(s)
- A. STENGEL
- CURE/Digestive Diseases Research Center, Center for Neurobiology of Stress, Department of Medicine, Digestive Diseases Division at the University of California Los Angeles, and VA Greater Los Angeles Health Care System, CA 90073, USA
| | - M. GOEBEL-STENGEL
- CURE/Digestive Diseases Research Center, Center for Neurobiology of Stress, Department of Medicine, Digestive Diseases Division at the University of California Los Angeles, and VA Greater Los Angeles Health Care System, CA 90073, USA
| | - L. WANG
- CURE/Digestive Diseases Research Center, Center for Neurobiology of Stress, Department of Medicine, Digestive Diseases Division at the University of California Los Angeles, and VA Greater Los Angeles Health Care System, CA 90073, USA
| | - A. LUCKEY
- CURE/Digestive Diseases Research Center, Center for Neurobiology of Stress, Department of Medicine, Digestive Diseases Division at the University of California Los Angeles, and VA Greater Los Angeles Health Care System, CA 90073, USA
| | - E. HU
- CURE/Digestive Diseases Research Center, Center for Neurobiology of Stress, Department of Medicine, Digestive Diseases Division at the University of California Los Angeles, and VA Greater Los Angeles Health Care System, CA 90073, USA
| | - J. RIVIER
- Peptide Biology Laboratories, Salk Institute, La Jolla, CA, USA
| | - Y. TACHÉ
- CURE/Digestive Diseases Research Center, Center for Neurobiology of Stress, Department of Medicine, Digestive Diseases Division at the University of California Los Angeles, and VA Greater Los Angeles Health Care System, CA 90073, USA
| |
Collapse
|
24
|
Koscielny A, Engel D, Maurer J, Hirner A, Kurts C, Kalff JC. Impact of CCR7 on the gastrointestinal field effect. Am J Physiol Gastrointest Liver Physiol 2011; 300:G665-75. [PMID: 21292999 DOI: 10.1152/ajpgi.00224.2010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Standardized intestinal manipulation (IM) leads to local bowel wall inflammation subsequently spreading over the entire gastrointestinal tract. Previously, we demonstrated that this so-called gastrointestinal field effect (FE) is immune mediated. This study aimed to investigate the role of CCR7 in IM-induced FE. Since CCR7 is expressed on activated dendritic cells and T cells and is well known to control their migration, we hypothesized that lack of CCR7 reduces or abolishes FE. Small bowel muscularis and colonic muscularis from CCR7(-/-) and wild-type (WT) mice were obtained after IM of the jejunum or sham operation. FE was analyzed by measuring gastrointestinal transit time of orally given fluorescent dextran (geometric center), colonic transit time, infiltration of MPO-positive cells, and circular smooth muscle contractility. Furthermore, mRNA levels of the inflammatory cytokine IL-6 were determined by RT-PCR. The number of dendritic cells and CD3+CD25+ T cells separately isolated from jejunum and colon was determined in mice after IM and sham operation. There was no significant difference in IL-6 mRNA upregulation in colonic muscularis between sham-operated WT and CCR7(-/-) mice after IM. Contractility of circular muscularis strips of the colon was significantly improved in CCR7(-/-) animals following IM and did not vary significantly from sham-operated animals. Additionally, inflammation of the colon determined by the number of MPO-positive cells and colonic transit time was significantly reduced in CCR7(-/-) mice. In contrast, jejunal contractility and jejunal inflammation of transgenic mice did not differ significantly from WT mice after IM. These data are supported by a significant increase of CD3+CD25+ T cells in the colonic muscularis of WT mice after IM, which could not be observed in CCR7(-/-) mice. These data demonstrate that CCR7 is required for FE and postoperative ileus. CCR7 indirectly affects FE by inhibiting migration of activated dendritic cells and of T cells from the jejunum to the colon. These findings support the critical role of the adaptive immune system in FE.
Collapse
Affiliation(s)
- A Koscielny
- Department of Surgery, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany.
| | | | | | | | | | | |
Collapse
|
25
|
Lubbers T, Buurman W, Luyer M. Controlling postoperative ileus by vagal activation. World J Gastroenterol 2010; 16:1683-1687. [PMID: 20379998 PMCID: PMC2852814 DOI: 10.3748/wjg.v16.i14.1683] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2009] [Revised: 01/25/2010] [Accepted: 02/01/2010] [Indexed: 02/06/2023] Open
Abstract
Postoperative ileus is a frequently occurring surgical complication, leading to increased morbidity and hospital stay. Abdominal surgical interventions are known to result in a protracted cessation of bowel movement. Activation of inhibitory neural pathways by nociceptive stimuli leads to an inhibition of propulsive activity, which resolves shortly after closure of the abdomen. The subsequent formation of an inflammatory infiltrate in the muscular layers of the intestine results in a more prolonged phase of ileus. Over the last decade, clinical strategies focusing on reduction of surgical stress and promoting postoperative recovery have improved the course of postoperative ileus. Additionally, recent experimental evidence implicated antiinflammatory interventions, such as vagal stimulation, as potential targets to treat postoperative ileus and reduce the period of intestinal hypomotility. Activation of nicotinic receptors on inflammatory cells by vagal input attenuates inflammation and promotes gastrointestinal motility in experimental models of ileus. A novel physiological intervention to activate this neuroimmune pathway is enteral administration of lipid-rich nutrition. Perioperative administration of lipid-rich nutrition reduced manipulation-induced local inflammation of the intestine and accelerated recovery of bowel movement. The application of safe and easy to use antiinflammatory interventions, together with the current multimodal approach, could reduce postoperative ileus to an absolute minimum and shorten hospital stay.
Collapse
|
26
|
Abstract
Constipation is a significant problem related to opioid medications used to manage pain. This review attempts to outline the latest findings related to the therapeutic usefulness of a μ opioid receptor antagonist, methylnaltrexone in the treatment of opioid-induced constipation. The review highlights methylnaltrexone bromide (Relistor™; Progenics/Wyeth) a quaternary derivative of naltrexone, which was recently approved in the United States, Europe and Canada. The Food and Drug Administration in the United States approved a subcutaneous injection for the treatment of opioid bowel dysfunction in patients with advanced illness who are receiving palliative care and when laxative therapy has been insufficient. Methylnaltrexone is a peripherally restricted, μ opioid receptor antagonist that accelerates oral-cecal transit in patients with opioid-induced constipation without reversing the analgesic effects of morphine or inducing symptoms of opioid withdrawal. An analysis of the mechanism of action and the potential benefits of using methylnaltrexone is based on data from published basic research and recent clinical studies.
Collapse
Affiliation(s)
- Beverley Greenwood-Van Meerveld
- Veterans Affairs Medical Center, Oklahoma Center for Neuroscience, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kelly M Standifer
- Department of Pharmaceutical Sciences, Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
27
|
Fraser GL, Venkova K, Hoveyda HR, Thomas H, Greenwood-Van Meerveld B. Effect of the ghrelin receptor agonist TZP-101 on colonic transit in a rat model of postoperative ileus. Eur J Pharmacol 2008; 604:132-7. [PMID: 19121631 DOI: 10.1016/j.ejphar.2008.12.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2008] [Revised: 11/21/2008] [Accepted: 12/09/2008] [Indexed: 12/26/2022]
Abstract
Ghrelin, the natural ligand of the growth hormone secretagogue receptor (ghrelin receptor), is an orexigenic gut hormone with prokinetic action in the upper gastrointestinal tract. Previously we have shown in a rodent model of postoperative ileus that the synthetic ghrelin receptor agonist TZP-101 prevents the delay in gastric emptying and improves small intestinal transit. The goal of the present study was to investigate whether TZP-101 affects colonic transit and food intake in rats with postoperative ileus. Fasted rats were treated with morphine and subjected to laparotomy under isoflurane anesthesia. Following surgery the animals were placed in clean home cages and fecal pellet output and food intake were monitored for 48 h. TZP-101 or vehicle were administered as 3 i.v. bolus infusions at 0 h, 2 h and 4 h post-surgery. TZP-101 (0.03-1 mg/kg) dose-dependently decreased the time to first bowel movement and increased fecal pellet output measured at 12 h and 24 h post-surgery compared to the vehicle. The administration of TZP-101 was not associated with a significant alteration in food intake. In conclusion, this study provides the first experimental evidence that a novel ghrelin receptor agonist improves large bowel function in rats with postoperative ileus, suggesting that TZP-101 may be useful in the clinic to accelerate upper gastrointestinal transit and to shorten the time to the first bowel movement following surgery.
Collapse
|
28
|
Vanneste G, Van Nassauw L, Kalfin R, Van Colen I, Elinck E, Van Crombruggen K, Timmermans JP, Lefebvre RA. Jejunal cholinergic, nitrergic, and soluble guanylate cyclase activity in postoperative ileus. Surgery 2008; 144:410-26. [PMID: 18707040 DOI: 10.1016/j.surg.2008.05.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2007] [Accepted: 05/17/2008] [Indexed: 01/15/2023]
Abstract
BACKGROUND In animal models of postoperative ileus (POI), inflammation of the intestine plays an important role in the pathogenesis of POI. Changes in alpha(2)-adrenoceptors and nitrergic regulation have been proposed to be implicated. The aim of our study was to investigate the presynaptic alpha(2)-receptor-mediated control of cholinergic nerve activity, the nitrergic nerve activity, and the possible role of soluble guanylate cyclase (sGC) during the inflammatory phase of POI. METHODS Ileus was induced by anesthesia and manipulation of the rat jejunum. Rats were treated with the sGC inhibitors methylene blue or ODQ; nonoperated animals served as controls. After 24 h, plasma and jejunal tissue were collected for biochemical assays, nitric oxide synthase-1 (NOS-1)-immunohistochemistry, acetylcholine (Ach)-release experiments, and muscle tension experiments. RESULTS In all operated animal groups, myeloperoxidase activity was significantly increased, which indicates initiation of an inflammatory response. The alpha(2)-adrenoceptor agonist UK14,304 reduced electrically induced Ach-release similarly in operated and nonoperated animals. In strips of operated animals, electrically induced nitrergic relaxations were decreased, whereas relaxations induced by exogenous nitric oxide (NO) remained unchanged compared with control. The number of myenteric neurons and the percentage of NOS-1-positive neurons were not influenced. Plasmatic cyclic guanosine monophosphate (cGMP) levels were decreased in all operated groups, whereas jejunal cGMP levels were unchanged compared with nonoperated controls; treatment with sGC inhibitors did not reduce plasmatic cGMP levels. CONCLUSIONS This study demonstrates that presynaptic alpha(2)-receptor mediated control of intestinal cholinergic nerve activity is unchanged during manipulation-induced inflammation. However, this inflammation induces impaired nitrergic neurotransmission related to decreased NOS-1 activity in the nitrergic nerves.
Collapse
Affiliation(s)
- Gwen Vanneste
- Heymans Institute of Pharmacology, Ghent University, Ghent, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
29
|
|
30
|
Fukuda H, Tsuchida D, Koda K, Miyazaki M, Pappas TN, Takahashi T. Inhibition of sympathetic pathways restores postoperative ileus in the upper and lower gastrointestinal tract. J Gastroenterol Hepatol 2007; 22:1293-9. [PMID: 17688668 DOI: 10.1111/j.1440-1746.2007.04915.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIM Postoperative ileus (POI) is a transient bowel dysmotility following abdominal surgery. The effects of adrenergic blocking agents and celiac ganglionectomy were studied in rats to investigate the possible involvement of the adrenergic pathway in whole gastrointestinal (GI) transit in the early and late phases of POI. METHODS After laparotomy, the terminal ileum was manipulated for 10 min. (51)Cr was administered into the stomach or colon immediately after surgery. In another group, (51)Cr was administered 24 h after surgery. Three hours after (51)Cr was administered, the rats were killed, and GI and colonic transit was calculated as a geometric center (GC). RESULTS Gastrointestinal transit was significantly delayed 3 h after intestinal manipulation, compared with GI transit in rats that had anesthesia only. Three hours after intestinal manipulation, guanethidine (5 mg/kg) and yohimbine (3 mg/kg) significantly improved delayed GI transit. Celiac ganglionectomy also significantly improved delayed GI transit. Twenty-seven hours after intestinal manipulation, guanethidine, yohimbine and celiac ganglionectomy improved delayed GI transit induced by intestinal manipulation. Colonic transit was delayed 3 h after intestinal manipulation, and delayed colonic transit was partially restored within 27 h of intestinal manipulation. Guanethidine, yohimbine and celiac ganglionectomy improved delayed colonic transit 3 h and 27 h after intestinal manipulation. CONCLUSIONS Adrenoceptors activated by intestinal manipulation impair the motility of the entire GI tract in both the early and the late phase of POI.
Collapse
Affiliation(s)
- Hiroyuki Fukuda
- Department of Surgery, Duke University Medical Center and VA Medical Center, Durham, North Carolina, USA
| | | | | | | | | | | |
Collapse
|
31
|
Fukuda H, Suenaga K, Tsuchida D, Mantyh CR, Pappas TN, Hicks GA, Dehaven-Hudkins DL, Takahashi T. The selective mu opioid receptor antagonist, alvimopan, improves delayed GI transit of postoperative ileus in rats. Brain Res 2006; 1102:63-70. [PMID: 16797494 DOI: 10.1016/j.brainres.2006.02.092] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2005] [Revised: 02/23/2006] [Accepted: 02/24/2006] [Indexed: 11/21/2022]
Abstract
Postoperative ileus (POI) is often exacerbated by opioid analgesic use during and following surgery, since mu opioid receptor activation results in a further delay of gastrointestinal (GI) transit. The effects of alvimopan, a novel, selective, and peripherally acting mu opioid receptor antagonist, and the reference compound methylnaltrexone, upon POI were investigated in rats. Under isoflurane anesthesia, POI was induced by laparotomy with intestinal manipulation. Immediately after the surgery, the rats received (51)Cr by gavage. Three hours after the surgery, the rats were sacrificed and GI transit was estimated using the geometric center (GC) of (51)Cr. Alvimopan (0.1-3 mg/kg) or methylnaltrexone (100 mg/kg) were administered by gavage either before or after the surgery, with or without morphine administration (1 mg/kg). GI transit was delayed by intestinal manipulation (GC = 2.92 +/- 0.17). Alvimopan (1 and 3 mg/kg) significantly reversed this delayed GI transit when administered 45 min prior to surgery. However, the effects of alvimopan were less pronounced when administered following surgery. Morphine administration further delayed GI transit induced by intestinal manipulation (GC = 1.97 +/- 0.11). Under these conditions, alvimopan (1 and 3 mg/kg) also significantly improved delayed GI transit when administered before surgery. Methylnaltrexone was inactive under all experimental conditions. These data suggest that mu opioid receptors play a role in the pathogenesis of POI, and that the clinical benefit reported to be afforded by alvimopan may be in part mediated via inhibition of an endogenous opioid release as well as blockade of the unwanted GI actions of analgesic agents.
Collapse
Affiliation(s)
- Hiroyuki Fukuda
- Department of Surgery, Duke University Medical Center, Surgical Service 112, VA Medical Center, Durham, NC 27705, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Czimmer J, Million M, Taché Y. Urocortin 2 acts centrally to delay gastric emptying through sympathetic pathways while CRF and urocortin 1 inhibitory actions are vagal dependent in rats. Am J Physiol Gastrointest Liver Physiol 2006; 290:G511-8. [PMID: 16223946 DOI: 10.1152/ajpgi.00289.2005] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
We characterized the influence of the selective corticotropin-releasing factor 2 (CRF(2)) receptor agonist human urocortin 2 (Ucn 2), injected intracisternally, on gastric emptying and its mechanism of action compared with intracisternal CRF or urocortin (Ucn 1) in conscious rats. The methylcellulose phenol red solution was gavaged 20 min after peptide injection, and gastric emptying was measured 20 min later. The intracisternal injection of Ucn 2 (0.1 and 1 microg) and Ucn 1 (1 microg) decreased gastric emptying to 37.8 +/- 6.9%, 23.1 +/- 8.6%, and 21.6 +/- 5.9%, respectively, compared with 58.4 +/- 3.8% after intracisternal vehicle. At lower doses, Ucn 2 (0.03 microg) and Ucn 1 (0.1 microg) had no effect. The CRF(2) antagonist astressin(2)-B (3 microg ic) antagonized intracisternal Ucn 2 (0.1 microg) and CRF (0.3 microg)-induced inhibition of gastric emptying. Vagotomy enhanced intracisternal Ucn 2 (0.1 or 1 microg)-induced inhibition of gastric emptying compared with sham-operated group, whereas it blocked intracisternal CRF (1 microg) inhibitory action (45.5 +/- 8.4% vs. 9.7 +/- 9.7%). Sympathetic blockade by bretylium prevented intracisternal and intracerebroventricular Ucn 2-induced delayed gastric emptying, whereas it did not influence intravenous Ucn 2-, intracisternal CRF-, and intracisternal Ucn 1-induced inhibition of gastric emptying. Prazosin abolished the intracisternal Ucn 2 inhibitory effect, whereas yohimbine and propranolol did not. None of the pretreatments modified basal gastric emptying. These data indicate that intracisternal Ucn 2 induced a central CRF(2)-mediated inhibition of gastric emptying involving sympathetic alpha(1)-adrenergic mechanisms independent from the vagus contrasting with the vagal-dependent inhibitory actions of CRF and Ucn 1.
Collapse
Affiliation(s)
- József Czimmer
- CURE/Digestive Diseases Research Center and Center for Neurovisceral Sciences and Women's Health, Division of Digestive Diseases, Department of Medicine, University of California, Los Angeles, CA 90073, USA
| | | | | |
Collapse
|
33
|
Affiliation(s)
- Benjamin Person
- Department of Colorectal Surgery, Cleveland Clinic Florida, Weston, FL, USA
| | | |
Collapse
|
34
|
The FO, de Jonge WJ, Bennink RJ, van den Wijngaard RM, Boeckxstaens GE. The ICAM-1 antisense oligonucleotide ISIS-3082 prevents the development of postoperative ileus in mice. Br J Pharmacol 2005; 146:252-8. [PMID: 15997238 PMCID: PMC1576259 DOI: 10.1038/sj.bjp.0706303] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Intestinal manipulation (IM) during abdominal surgery triggers the influx of inflammatory cells, leading to postoperative ileus. Prevention of this local muscle inflammation, using intercellular adhesion molecule-1 (ICAM-1) and leukocyte function-associated antigen-1-specific antibodies, has been shown to shorten postoperative ileus. However, the therapeutic use of antibodies has considerable disadvantages. The aim of the current study was to evaluate the effect of ISIS-3082, a mouse-specific ICAM-1 antisense oligonucleotide, on postoperative ileus in mice. Mice underwent a laparotomy or a laparotomy combined with IM after treatment with ICAM-1 antibodies, 0.1-10 mg kg(-1) ISIS-3082, saline or ISIS-8997 (scrambled control antisense oligonucleotides, 1 and 3 mg kg(-1)). At 24 h after surgery, gastric emptying of a 99mTC labelled semi-liquid meal was determined using scintigraphy. Intestinal inflammation was assessed by myeloperoxidase (MPO) activity in ileal muscle whole mounts. IM significantly reduced gastric emptying compared to laparotomy. Pretreatment with ISIS-3082 (0.1-1 mg kg(-1)) as well as ICAM-1 antibodies (10 mg kg(-1)), but not ISIS-8997 or saline, improved gastric emptying in a dose-dependent manner. This effect diminished with higher doses of ISIS-3082 (3-10 mg kg(-1)). Similarly, ISIS-3082 (0.1-1 mg kg(-1)) and ICAM-1 antibodies, but not ISIS-8997 or higher doses of ISIS-3082 (3-10 mg kg(-1)), reduced manipulation-induced inflammation. Immunohistochemistry showed reduction of ICAM-1 expression with ISIS-3082 only. ISIS-3082 pretreatment prevents postoperative ileus in mice by reduction of manipulation-induced local intestinal muscle inflammation. Our data suggest that targeting ICAM-1 using antisense oligonucleotides may represent a new therapeutic approach to the prevention of postoperative ileus.
Collapse
Affiliation(s)
- Frans O The
- Department of Gastroenterology and Hepatology, Academical Medical Center, Meibergdreef 9, 1055 AZ, Amsterdam, The Netherlands
| | - Wouter J de Jonge
- Department of Gastroenterology and Hepatology, Academical Medical Center, Meibergdreef 9, 1055 AZ, Amsterdam, The Netherlands
| | - Roel J Bennink
- Department of Nuclear Medicine, Academical Medical Center, Amsterdam, The Netherlands
| | - Rene M van den Wijngaard
- Department of Gastroenterology and Hepatology, Academical Medical Center, Meibergdreef 9, 1055 AZ, Amsterdam, The Netherlands
| | - Guy E Boeckxstaens
- Department of Gastroenterology and Hepatology, Academical Medical Center, Meibergdreef 9, 1055 AZ, Amsterdam, The Netherlands
- Author for correspondence:
| |
Collapse
|
35
|
Fukuda H, Tsuchida D, Koda K, Miyazaki M, Pappas TN, Takahashi T. Impaired gastric motor activity after abdominal surgery in rats. Neurogastroenterol Motil 2005; 17:245-50. [PMID: 15787944 DOI: 10.1111/j.1365-2982.2004.00602.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Postoperative ileus (POI) is a transient bowel dysmotility that occurs following abdominal surgery. Several mechanisms have been proposed such as neural reflex and inflammatory changes. We focused on gastric motility after abdominal surgery in rats. To investigate the time course of gastric motility after surgery, gastric motility was continuously recorded before, during and after surgery. After laparotomy, terminal ileum was manipulated for 10 min. Gastric motility was recorded by a strain gauge transducer implanted on the serosal surface of the stomach. To investigate whether peripheral sympathetic nerve is involved in the pathogenesis of POI, effects of guanethidine and celiac ganglionectomy were tested on the postoperative gastric motility. Although isoflurane anaesthesia reduced the gastric motility to 40%, the motility recovered immediately when isoflurane was withdrawn. Intestinal manipulation reduced the postoperative gastric motility for 3-24 h after surgery, compared with preoperative levels. Guanethidine administration and celiac ganglionectomy restored the impaired gastric motility. Feeding increased the gastric motility in each group. It is suggested that the pathogenesis of postoperative gastric ileus induced by intestinal manipulation involves viscero-sympathetic pathways. Intestinal manipulation causes impaired gastric motility via inhibitory sympathetic efferent pathway. Feeding may improve the postoperative gastric motility.
Collapse
Affiliation(s)
- H Fukuda
- Department of Surgery, Duke University Medical Center and VA Medical Center, Durham, NC 27705, USA
| | | | | | | | | | | |
Collapse
|
36
|
de Winter BY, van Nassauw L, de Man JG, de Jonge F, Bredenoord AJ, Seerden TC, Herman AG, Timmermans JP, Pelckmans PA. Role of oxidative stress in the pathogenesis of septic ileus in mice. Neurogastroenterol Motil 2005; 17:251-261. [PMID: 15787945 DOI: 10.1111/j.1365-2982.2004.00618.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
We investigated the role of oxidative stress in the pathogenesis of septic ileus. Sepsis was induced by intraperitoneal (i.p.) injection of lipopolysaccharides (LPS, 20 mg kg(-1)) in mice. The effect of two i.p. injections of superoxide dismutase [polyethylene glycol (PEG)-SOD, 4000 U kg(-1)] and catalase (PEG-CAT, 15,000 U kg(-1)) was investigated on gastric emptying, intestinal transit and total nitrite plasma concentrations. We also performed immunohistochemical experiments on gastric and ileal tissue. LPS significantly delayed gastric emptying and intestinal transit while plasma nitrite levels increased. Polyethylene glycol (PEG)-SOD reversed the endotoxin-induced delay in gastric emptying and improved the delay in intestinal transit without effect on plasma nitrite levels. PEG-CAT slightly improved the delay in gastric emptying without effect on intestinal transit. Immunohistochemistry showed the presence of nitrotyrosine (NT) and 4-hydroxy-2-nonenal (HNE) in the gastric and ileal mucosa of LPS-treated mice. Treatment with PEG-SOD or PEG-CAT of LPS mice diminished the presence of NT or HNE in both tissues. In addition, LPS induced a significant increase in inducible nitric oxide synthase (iNOS)-positive residential macrophages in the external musculature of stomach and ileum, which significantly decreased after PEG-SOD or PEG-CAT treatment. The present results support a role for oxidative and nitrosative stress in the pathogenesis of septic ileus in mice.
Collapse
Affiliation(s)
- B Y de Winter
- Division of Gastroenterology, Faculty of Medicine, University of Antwerp (campus Drie Eiken), Universiteitsplein 1, 2610 Antwerp, Belgium.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Postoperative gastrointestinal (GI) tract dysfunction (PGID) is common and is associated with increased patient suffering and cost of care. The pathogenesis of PGID is complex and multifactorial. Traditional measures intended to reduce the incidence of PGID, such as the use of prokinetic drugs, nasogastric tube drainage, and the avoidance of early fluid and/or food intake, are apparently not beneficial. The administration of larger volumes of IV fluids to achieve predetermined increases in cardiac output has been shown in randomized trials to improve gut perfusion and reduce the incidence of PGID. A multimodal approach that includes limited surgical incision, regional local anesthesia, early mobilization, and enteral feeding has been associated with a dramatic reduction in postoperative complications, PGID, and length of hospital stay. However, none of these approaches has been validated in adequately powered multicenter prospective randomized controlled trials.
Collapse
Affiliation(s)
- Michael G Mythen
- Department of Anaesthesia and Critical Care, University College London, United Kingdom; and Portex Anaesthesia, Intensive Care and Respiratory Unit, Institute of Child Health, University College London, United Kingdom
| |
Collapse
|
38
|
Abstract
Postoperative ileus is an iatrogenic condition that follows abdominal surgery. Three main mechanisms are involved in its causation, namely neurogenic, inflammatory and pharmacological mechanisms. In the acute postoperative phase, mainly spinal and supraspinal adrenergic and non-adrenergic pathways are activated. Recent studies, however, show that the prolonged phase of postoperative ileus is caused by an enteric molecular inflammatory response and the subsequent recruitment of leucocytes into the muscularis of the intestinal segments manipulated during surgery. This inflammation impairs local neuromuscular function and activates neurogenic inhibitory pathways, inhibiting motility of the entire gastrointestinal tract. The mechanisms underlying the recruitment of the inflammatory cells, and their interaction with the intestinal afferent innervation, are discussed. Finally, opioids administered for postoperative pain control also contribute to a large extent to the reduction in propulsive gastrointestinal motility observed after abdominal surgery.
Collapse
Affiliation(s)
- A J Bauer
- Department of Medicine/Gastroenterology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | | |
Collapse
|
39
|
de Jonge WJ, van den Wijngaard RM, The FO, ter Beek ML, Bennink RJ, Tytgat GNJ, Buijs RM, Reitsma PH, van Deventer SJ, Boeckxstaens GE. Postoperative ileus is maintained by intestinal immune infiltrates that activate inhibitory neural pathways in mice. Gastroenterology 2003; 125:1137-47. [PMID: 14517797 DOI: 10.1016/s0016-5085(03)01197-1] [Citation(s) in RCA: 157] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND AND AIMS Postoperative ileus after abdominal surgery largely contributes to patient morbidity and prolongs hospitalization. We aimed to study its pathophysiology in a murine model by determining gastric emptying after manipulation of the small intestine. METHODS Gastric emptying was determined at 6, 12, 24, and 48 hours after abdominal surgery by using scintigraphic imaging. Intestinal or gastric inflammation was assessed by immune-histochemical staining and measurement of tissue myeloperoxidase activity. Neuromuscular function of gastric and intestinal muscle strips was determined in organ baths. RESULTS Intestinal manipulation resulted in delayed gastric emptying up to 48 hours after surgery; gastric half-emptying time 24 hours after surgery increased from 16.0 +/- 4.4 minutes after control laparotomy to 35.6 +/- 5.4 minutes after intestinal manipulation. The sustained delay in gastric emptying was associated with the appearance of leukocyte infiltrates in the muscularis of the manipulated intestine, but not in untouched stomach or colon. The delay in postoperative gastric emptying was prevented by inhibition of intestinal leukocyte recruitment. In addition, postoperative neural blockade with hexamethonium (1 mg/kg intraperitoneally) or guanethidine (50 mg/kg intraperitoneally) normalized gastric emptying without affecting small-intestinal transit. The appearance of intestinal infiltrates after intestinal manipulation was associated with increased c-fos protein expression in sensory neurons in the lumbar spinal cord. CONCLUSIONS Sustained postoperative gastroparesis after intestinal manipulation is mediated by an inhibitory enterogastric neural pathway that is triggered by inflammatory infiltrates recruited to the intestinal muscularis. These findings show new targets to shorten the duration of postoperative ileus pharmacologically.
Collapse
Affiliation(s)
- Wouter J de Jonge
- Department of Gastroenterology and Hepatology, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Luckey A, Wang L, Jamieson PM, Basa NR, Million M, Czimmer J, Vale W, Taché Y. Corticotropin-releasing factor receptor 1-deficient mice do not develop postoperative gastric ileus. Gastroenterology 2003; 125:654-9. [PMID: 12949710 DOI: 10.1016/s0016-5085(03)01069-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Corticotropin-releasing factor (CRF) signaling pathways play a key role in the stress response through the activation of CRF(1) and CRF(2) receptors. We investigated the CRF receptor subtypes involved in gastric postoperative ileus. METHODS Adult male mice (C57BL/6, CRF(1)-deficient, and wild-type), fasted for 16-18 hours, were anesthetized for 10 minutes and had a midline celiotomy and cecal exteriorization and palpation for 30 or 60 seconds or no surgery (sham). Phenol red was given by gavage 100 minutes after anesthesia; 20 minutes later, gastric emptying and blood glucose level were measured. RESULTS In C57BL/6 mice, cecal palpation for 30 or 60 seconds significantly reduced gastric emptying to 30.3% +/- 1.4% and 5.8% +/- 3.4%, respectively, compared with 58.5% +/- 4.4% in sham. The CRF(1) antagonist CP-154,526 (20 mg/kg subcutaneously) completely prevented the 30-second cecal palpation-induced delayed gastric emptying (53.0% +/- 7.9% vs. 28.0% +/- 4.0% in vehicle + surgery), whereas the CRF(2) antagonist astressin(2)-B injected subcutaneously had no effect. In CRF(1)-deficient mice, cecal palpation for 30 seconds did not delay gastric emptying (80.3% +/- 4.5% compared with 84.7% +/- 6.3% in sham); in wild-type mice, gastric emptying was decreased to 17.8% +/- 16.1% (P < 0.05 vs. sham 72.0% +/- 12.4%). Surgery increased glucose levels by 46% compared with sham in wild-type mice, while glycemia was not altered in CRF(1)-deficient mice. Basal emptying was similar in wild-type and CRF(1)-deficient mice and not influenced by CRF antagonists in C57BL/6 mice. CONCLUSIONS These data show that CRF(1) activation plays an important role in mediating the early phase of gastric ileus.
Collapse
Affiliation(s)
- Andrew Luckey
- Department of Veternas Affairs, Greater Los Angeles Healthcare System, La Jolla, California 90073, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Berthoud HR, Hennig G, Campbell M, Volaufova J, Costa M. Video-based spatio-temporal maps for analysis of gastric motility in vitro: effects of vagal stimulation in guinea-pigs. Neurogastroenterol Motil 2002; 14:677-88. [PMID: 12464090 DOI: 10.1046/j.1365-2982.2002.00369.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Our aim was to evaluate topographically specific gastric motility changes induced by graded vagal activation. A recently developed method of constructing spatio-temporal maps of motility from video movies was adapted to the in vitro perfused guinea-pig stomach with an intact vagal nerve supply. In the unstimulated preparation, spontaneous activity was low or absent. Bilateral vagal stimulation with frequencies as low as 0.2 Hz triggered weak anally, and in some cases orally, propagating antral contractions at rates of about 5-6 min-1. Upon stimulation with higher frequencies, antral contractions increased significantly in length (starting more proximally) and amplitude, and produced large pressure peaks of up to 25 hPa, with maximal effects at 2-4 Hz. In contrast, the speed of propagation and the interval between peristaltic waves did not change with vagal stimulation at any frequency. Vagal stimulation also produced a significant and frequency-dependent enlargement of the fundus with a maximal effect at 4 Hz. It is concluded that a very low tonic vagal activity is apparently necessary and sufficient to express basic antral motility, while more sustained vagal activity is necessary for high-amplitude gastric contractions and significant sustained fundic relaxation. The constant interval between propagating contractions supports the concept that vagal input impinges on intrinsic enteric neural circuits that have a modulatory role in the myogenic mechanism underlying slow-wave peristalsis, rather than directly on gastric musculature.
Collapse
Affiliation(s)
- H-R Berthoud
- Neurobiology of Nutrition Laboratory, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge 70808, USA.
| | | | | | | | | |
Collapse
|
42
|
Bauer AJ, Schwarz NT, Moore BA, Türler A, Kalff JC. Ileus in critical illness: mechanisms and management. Curr Opin Crit Care 2002; 8:152-7. [PMID: 12386517 DOI: 10.1097/00075198-200204000-00011] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nonobstructive ileus, signifying the impairment of coordinated propulsive intestinal motility, remains a frequently documented and almost inevitable consequence of open abdominal surgery and sepsis. Despite the frequency and major impact of ileus on morbidity and mortality, the exact underlying molecular and cellular mechanisms of this important clinical conundrum are still ill defined. Animal models suggest that both neuronal and local inflammatory responses within the intestinal muscularis mechanistically contribute to intestinal ileus. The neuronal mechanism appears to involve the enhanced release of nitric oxide from inhibitory motor neurons. Likewise, nitric oxide and prostaglandins are released from inflammatory cells (macrophages and monocytes) via the induction of nitric oxide synthase (iNOS) and cyclooxygenase-2. Recently, preliminary data have confirmed the existence of an intraoperative local muscularis inflammatory response during surgery in human patients.
Collapse
Affiliation(s)
- Anthony J Bauer
- Department of Medicine/Gastroenterology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania 15216, USA. tbauer+@pitt.edu
| | | | | | | | | |
Collapse
|
43
|
Boeckxstaens GE, Hollmann M, Heisterkamp SH, Robberecht P, de Jonge WJ, van Den Wijngaard RM, Tytgat GN, Blommaart PJ. Evidence for VIP(1)/PACAP receptors in the afferent pathway mediating surgery-induced fundic relaxation in the rat. Br J Pharmacol 2000; 131:705-10. [PMID: 11030719 PMCID: PMC1572383 DOI: 10.1038/sj.bjp.0703625] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
We previously reported activation of an inhibitory adrenergic and a non-adrenergic non-cholinergic (NANC) pathway during abdominal surgery relaxing the rat gastric fundus. In the present study, we investigated the possible role of nitric oxide (NO) and vasoactive intestinal polypeptide (VIP) in the NANC part of the surgery-induced fundic relaxation. The effect of the NO biosynthesis inhibitor N(G)-nitro-L-arginine (L-NOARG), the non-selective VIP receptor antagonist [D-p-Cl-Phe(6),Leu(17)]-VIP and the selective VIP(1) receptor antagonist [Acetyl-His(1),D-Phe(2),Lys(15),Arg(16), Leu(17)]-VIP was investigated on the non-adrenergic fundic relaxation induced by manipulation of the small intestine followed by resection of the caecum. Guanethidine partly reduced the manipulation-induced fundic relaxation. Addition of L-NOARG reduced this non-adrenergic component, whereas the non-selective VIP receptor antagonist had no significant effect. Combination of L-NOARG and the non-selective VIP antagonist however further reduced the relaxation to manipulation. The selective VIP(1) receptor antagonist reduced the mean and maximal relaxation induced by abdominal surgery in the presence of guanethidine. When combined with L-NOARG, the relaxation of the gastric fundus was almost completely abolished. The VIP(1) receptor antagonist alone had no significant effect on the mean and maximal relaxation, but enhanced recovery of fundic tone. In conclusion, as VIP(1) receptors are not present in the rat gastric fundus, these results suggest that the NANC inhibitory pathway activated during abdominal surgery involves VIP(1) receptors, most likely in the afferent limb. The inhibitory neurotransmitters released at the level of the gastric fundus smooth muscle are NO and a substance different from VIP.
Collapse
Affiliation(s)
- G E Boeckxstaens
- Division of Gastroenterology & Hepatology, Academic Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|